Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Dusan Popadic is active.

Publication


Featured researches published by Dusan Popadic.


Glia | 2004

Astrocyte-induced regulatory T cells mitigate CNS autoimmunity.

Vladimir Trajkovic; O. Vuckovic; Stanislava Stosic-Grujicic; Djordje Miljković; Dusan Popadic; Milos Markovic; Vladimir Bumbasirevic; Aleksandar Backovic; Ivana Cvetkovic; Ljubica Harhaji; Zorica Ramić; Marija Mostarica Stojković

Although astrocytes presumably participate in maintaining the immune privilege of the central nervous system (CNS), the mechanisms behind their immunoregulatory properties are still largely undefined. In this study, we describe the development of regulatory T cells upon contact with astrocytes. Rat T cells pre‐incubated with astrocytes completely lost the ability to proliferate in response to mitogenic stimuli. The cells were blocked in G0/G1 phase of the cell cycle, expressed less IL‐2R, and produced significantly lower amounts of interferon‐γ (IFN‐γ), but not interleukin‐2 (IL‐2), IL‐10, or tumor necrosis factor (TNF). These anergic cells completely prevented mitogen‐induced growth of normal T lymphocytes, as well as CNS antigen‐driven proliferation of autoreactive T cells. The suppressive activity resided in both CD4+ and CD8+ T‐cell compartments. Heat‐sensitive soluble T‐cell factors, not including transforming growth factor‐β (TGF‐β) or IL‐10, were solely responsible for the observed suppression, as well as for the transfer of suppressive activity to normal T cells. The administration of astrocyte‐induced regulatory T cells markedly alleviated CNS inflammation and clinical symptoms of CNS autoimmunity in rats with experimental allergic encephalomyelitis. Finally, the cells with suppressive properties were readily generated from human lymphocytes after contact with astrocytes. Taken together, these data indicate that astrocyte‐induced regulatory T cells might represent an important mechanism for self‐limitation of excessive inflammation in the brain.


Journal of Cellular Physiology | 2008

Macrophage migration inhibitory factor (MIF) is necessary for progression of autoimmune diabetes mellitus

Stanislava Stosic-Grujicic; Ivana Stojanovic; Danijela Maksimovic-Ivanic; Miljana Momčilović; Dusan Popadic; Ljubica Harhaji; Djordje Miljković; Christine N. Metz; Katia Mangano; Gianpaolo Papaccio; Yousef Al-Abed; Ferdinando Nicoletti

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine of the innate immune system that plays a major role in the induction of immunoinflammatory responses. To examine the role of endogenous MIF in the pathogenesis of type 1 diabetes (TID) we evaluated the effects of administration of neutralizing anti‐MIF antibodies to NOD mice with accelerated forms of diabetes induced by injection of cyclophosphamide or by transfer of diabetogenic spleen cells. Both accelerated forms of diabetes were markedly reduced by anti‐MIF antibody. Furthermore, MIF‐deficient (MIF−/−) mice were less susceptible to the induction of immunoinflammatory diabetes, insulitis and apoptosis within the endocrine pancreas by multiple low doses of streptozotocin (MLD‐STZ) than genetically matched wild type (WT) mice. MIF deficiency resulted in lower proliferation and lymphocyte adhesion, as well as reduced production from the spleens and peritoneal cells of a variety of inflammatory mediators typically associated with development of the disease including IL‐12, IL‐23, TNF‐α, and IL‐1β. Furthermore, MIF deletion affected the production of IL‐18, TNF‐α, IL‐1β, and iNOS in the islets of Langerhans. These data, along with the higher expression of IL‐4 and TGF‐β observed in the periphery and in the pancreas of MLD‐STZ‐challenged MIF−/− mice as compared to WT controls suggest that MIF deficiency has induced an immune deviation towards protective type 2/3 response. These results suggest that MIF participates in T1D by controlling the functional activity of monocytes/macrophages and T cells and modulating their secretory capacity of pro‐ and anti‐inflammatory molecules. J. Cell. Physiol. 215: 665–675, 2008.


Journal of Neuroscience Research | 2006

Strain difference in susceptibility to experimental autoimmune encephalomyelitis between Albino Oxford and Dark Agouti rats correlates with disparity in production of IL-17, but not nitric oxide

Djordje Miljković; Stanislava Stosic-Grujicic; Milos Markovic; Miljana Momčilović; Zorica Ramić; Danijela Maksimovic-Ivanic; Sanja Mijatović; Dusan Popadic; Ivana Cvetkovic; Marija Mostarica-Stojkovic

Albino Oxford (AO) rats, unlike Dark Agouti (DA) rats are resistant to the induction of experimental autoimmune encephalomyelitis (EAE). The reason for the resistance could be some restraining mechanism preventing auto‐aggressive cell activation at the level of draining lymph nodes (DLN) during the induction phase of the disease. Such a mechanism could be anti‐proliferative action of nitric oxide (NO), which has already been shown of importance for the resistance of several rat strains to the induction of the disease. Importantly, number of AO DLN cells (DLNC) is markedly lower and with lower proliferative response to myelin basic protein (MBP) ex vivo in comparison to DA DLNC in the inductive phase of EAE, thus implying that in AO rats DLNC do not proliferate as extensively as in DA rats. We show that AO rats do not produce larger quantities of NO than DA rats after immunization. Further, DLNC of immunized AO rats have significantly lower mRNA expression and synthesis of interferon (IFN)‐γ and interleukin (IL)‐17 compared to DLNC of DA rats. Collectively, these results suggest that there is a substantial difference between EAE‐resistant AO rats and EAE‐prone DA rats in the initiation of autoimmune response. This difference seems to be independent of anti‐proliferative actions of NO, but correlates with impaired IL‐17 production in AO rats.


ChemMedChem | 2010

Synthesis and in vitro Anticancer Activity of Octahedral Platinum(IV) Complexes with Cyclohexyl‐Functionalized Ethylenediamine‐N,N′‐Diacetate‐Type Ligands

Jelena Lazic; Ljubica Vucicevic; Sanja Grgurić-Šipka; Kristina Janjetovic; Goran N. Kaluđerović; Maja Misirkic; Maja Gruden-Pavlović; Dusan Popadic; Reinhard Paschke; Vladimir Trajkovic; Tibor J. Sabo

The present study describes the synthesis and anticancer activity of novel octahedral PtIV complexes with cyclohexyl functionalized ethylenediamine‐N,N′‐diacetate‐type ligands. Molecular mechanics calculations and density functional theory analysis revealed that s‐cis is the preferred geometry of these PtIV complexes with tetradentate‐coordinated (S,S)‐ethylenediamine‐N,N′‐di‐2‐(3‐cyclohexyl)propanoate. The viability of cancer cell lines (U251 human glioma, C6 rat glioma, L929 mouse fibrosarcoma, and B16 human melanoma) was assessed by measuring mitochondrial dehydrogenase activity and lactate dehydrogenase release. Cell‐cycle distribution, oxidative stress, caspase activation, and induction of autophagy were analyzed by flow cytometry using appropriate fluorescent reporter dyes. The cytotoxic activity of novel PtIV complexes against various cancer cell lines (IC50 range: 1.9–8.7 μM) was higher than that of cisplatin (IC50 range: 10.9–67.0 μM) and proceeded through completely different mechanisms. Cisplatin induced caspase‐dependent apoptosis associated with the cytoprotective autophagic response. In contrast, the new PtIV complexes caused rapid, caspase‐independent, oxidative stress‐mediated non‐apoptotic cell death characterized by massive cytoplasmic vacuolization, cell membrane damage, and the absence of protective autophagy.


Brain Research | 2001

Leflunomide inhibits activation of inducible nitric oxide synthase in rat astrocytes.

Dj. Miljkovic; Tatjana Samardzic; M. Mostarica Stojkovic; S. Stosic-Grujicic; Dusan Popadic; Vladimir Trajkovic

Highly reactive gaseous free radical nitric oxide (NO), generated by astrocytes and infiltrating macrophages is implicated in inflammatory destruction of brain tissue, including that occurring in multiple sclerosis. Therefore, the influence of immunosuppressive drug leflunomide on inducible nitric oxide synthase (iNOS)-dependent NO production in rat astrocytes and macrophages was investigated. Under the same cultivating conditions, leflunomides active metabolite A77 1726 caused a dose-dependent decrease of NO production in IFN-gamma+LPS-stimulated primary astrocytes, but not in macrophages. While A77 1726 did not alter iNOS enzymatic activity, it markedly suppressed IFN-gamma+LPS-triggered expression of iNOS mRNA in astrocytes. In the presence of transcription inhibitor actinomycin D, A77 1726 failed to inhibit astrocyte NO production, suggesting transcriptional regulation of iNOS by leflunomide. This assumption was further supported by the ability of A77 1726 to inhibit IFN-gamma+LPS-induced expression of mRNA for an important iNOS transcription factor IRF-1. PD98059, a specific inhibitor of mitogen-activated protein kinase kinase (MAPKK/MEK), but not genistein, an unselective protein tyrosine kinase inhibitor, completely mimicked cell type-specific inhibition of NO synthesis by A77 1726. Therefore, previously described inhibition of MEK/MAP pathway by leflunomide could present a possible mechanism for A77 1726-mediated suppression of iNOS activation in astrocytes. Accordingly to results obtained with primary astrocytes, both A77 1726 and PD98059 significantly reduced IFN-gamma+LPS-induced NO synthesis in the cultures of rat astrocytoma cell line C6. The ability to suppress iNOS induction in astrocytes supports potential use of leflunomide in the treatment of multiple sclerosis and other NO-dependent inflammatory brain disorders.


Cellular and Molecular Life Sciences | 2004

Aloe-emodin prevents cytokine-induced tumor cell death: the inhibition of auto-toxic nitric oxide release as a potential mechanism.

S. Mijatovic; D. Maksimovic-Ivanic; J. Radovic; Dusan Popadic; M. Momcilovic; Lj. Harhaji; Dj. Miljkovic; Vladimir Trajkovic

AbstractAloe-emodin (AE) is a plant-derived hydroxyanthraquinone with potential anticancer activity. We investigated the ability of AE to modulate survival of mouse L929 fibrosarcoma and rat C6 astrocytoma cells through interference with the activation of inducible nitric oxide (NO) synthase (NOS) and subsequent production of tumoricidal free radical NO. Somewhat surprisingly, AE in a dose-dependent manner rescued inter-feron-γ + interleukin-1-stimulated L929 cells from NO-dependent killing by reducing their autotoxic NO release. The observed protective effect was less pronounced in C6 cells, due to their higher sensitivity to a direct toxic action of the drug. AE-mediated inhibition of tumor cell NO release coincided with a reduction in cytokine-induced accumulation of transcription and translation products of genes encoding inducible NOS and its transcription factor IRF-1, while activation of NF-κB remained unaltered. These data indicate that the influence of AE on tumor growth might be more complex that previously recognized, the net effect being determined by the balance between the two opposing actions of the drug: its capacity to directly kill tumor cells, but also to protect them from NO-mediated toxicity.


Journal of Neuroimmunology | 2009

Expression of Th1 and Th17 cytokines and transcription factors in multiple sclerosis patients: does baseline T-bet mRNA predict the response to interferon-beta treatment?

Jelena Drulovic; Emina Savic; Tatjana Pekmezovic; Sarlota Mesaros; Nebojsa Stojsavljevic; Irena Dujmovic-Basuroski; Jelena Kostic; Vladimir Vasic; Marija Mostarica Stojković; Dusan Popadic

We studied the effect of one-year interferon (IFN)-beta treatment on the in vivo mRNA expression of IFN-gamma, interleukin (IL)-17, T-bet and RoR-gammat, on peripheral blood mononuclear cells (PBMC) from 36 multiple sclerosis (MS) patients. In the total MS group, IFN-beta induced decrease in mRNA levels of IFN-gamma and T-bet (p<0.0001), while the levels of IL-17 and RoR-gammat remained similar. In both responders and non-responders, IFN-beta induced significant decrease of IFN-gamma (p<0.0001 and p=0.011, respectively), while decrease in T-bet was detected only in responders (p<0.0001). Higher pre-treatment T-bet allowed prediction of the clinical response in the first year (beta=0.601, p=0.036). Our preliminary findings suggest that T-bet expression might be a potential prognostic marker of treatment response to IFN-beta in MS.


Immunology | 1997

Cell-specific effects of pentoxifylline on nitric oxide production and inducible nitric oxide synthase mRNA expression.

Vladimir Trajkovic; Vladimir P. Badovinac; Dusan Popadic; O Hadzić; M. Mostarica Stojkovic

Cytokine‐stimulated astrocytes and macrophages are potent producers of nitric oxide (NO), a free radical proposed to play an important role in organ‐specific autoimmunity, including demyelinating diseases of the central nervous system. The aim of this study was to investigate effects of pentoxifylline (PTX), a phosphodiesterase inhibitor with immunomodulatory properties, on NO production and inducible NO synthase (iNOS) mRNA expression in rat astrocytes and macrophages. We have shown that PTX affects cytokine (interferon‐γ, IFN‐γ; interleukin‐1, IL‐1; tumour‐necrosis factor‐α, TNF‐α)‐induced NO production in both cell types, but in the opposite manner – enhancing in astrocytes and suppressive in macrophages. While PTX did not have any effect on enzymatic activity of iNOS in activated cells, expression of iNOS mRNA was elevated in astrocytes and decreased in macrophages treated with cytokines and PTX. Treatment with PTX alone affected neither NO production nor iNOS mRNA levels in astrocytes or macrophages. This study indicates involvement of different signalling pathways associated with iNOS induction in astrocytes and macrophages, thus emphasizing complexity of regulation of NO synthesis in different cell types.


Neuroscience Letters | 2008

Kinetics of IFN-γ and IL-17 expression and production in active experimental autoimmune encephalomyelitis in Dark Agouti rats

Miljana Momčilović; Željka Miljković; Dusan Popadic; Djordje Miljković; Marija Mostarica-Stojkovic

Interferon-gamma (IFN-gamma) and interleukin-17 (IL-17) have been involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). We have carried out a follow-up study of the expression and production of these cytokines, as well as of cells expressing these cytokines during the course of active EAE in Dark Agouti (DA) rats. As a result, IL-17, but not IFN-gamma expression and production had the peak value in draining lymph nodes (DLN) during the induction phase of the disease, and in spinal cords (SC) at the onset of clinical signs of the disease, and then declined toward the resolution of the disease. Also, a significant proportion of IFN-gamma/IL-17 double-positive cells was observed in SC of DA rats in active EAE. Importantly, the highest proportion of IL-17 single positive and double-positive cells, but not of IFN-gamma single positive cells, was observed at the onset of the disease. The observed difference in the kinetics of IFN-gamma and IL-17 expression during active EAE in DA rats suggests different roles these cytokines might have in the pathogenesis of the disease.


Journal of Pharmacology and Experimental Therapeutics | 2006

A Potent Immunomodulatory Compound, (S,R)-3-Phenyl-4,5-dihydro-5-isoxasole Acetic Acid, Prevents Spontaneous and Accelerated Forms of Autoimmune Diabetes in NOD Mice and Inhibits the Immunoinflammatory Diabetes Induced by Multiple Low Doses of Streptozotocin in CBA/H Mice

Stanislava Stosic-Grujicic; Ivana Cvetkovic; Katia Mangano; Massimo Fresta; Danijela Maksimovic-Ivanic; Ljubica Harhaji; Dusan Popadic; Miljana Momčilović; Djordje Miljković; Joseph A. Kim; Yousef Al-Abed; Ferdinando Nicoletti

(S,R)-3-Phenyl-4,5-dihydro-5-isoxasole acetic acid (VGX-1027) is an isoxazole compound that exhibits various immunomodulatory properties. The capacity of VGX-1027 to prevent interleukin (IL)-1β plus interferon-γ-induced pancreatic islet death in vitro prompted us to evaluate its effects on the development of autoimmune diabetes in preclinical models of human type 1 diabetes mellitus (T1D). Administration of VGX-1027 to NOD mice with spontaneous or accelerated forms of diabetes induced either by injection of cyclophosphamide or by transfer of spleen cells from acutely diabetic syngeneic donors markedly reduced the cumulative incidence of diabetes and insulitis. In addition, VGX-1027 given either i.p. or p.o. to CBA/H mice made diabetic with multiple low doses of streptozotocin successfully counteracted the development of destructive insulitis and hyperglycemia. The animals receiving VGX-1027 exhibited reduced production of the proinflammatory mediators tumor necrosis factor-α, IL-1β, macrophage migration inhibitory factor, and inducible nitric-oxide synthase-mediated nitric oxide generation in both pancreatic islets and peripheral compartments. These results indicate that VGX-1027 probably exerts its antidiabetogenic effects by limiting cytokine-mediated immunoinflammatory events, leading to inflammation and destruction of pancreatic islets. VGX-1027 seems worthy of being considered as a candidate drug in the development of new therapeutic strategies for the prevention and early treatment of T1D.

Collaboration


Dive into the Dusan Popadic's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emina Savic

University of Belgrade

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge